Uveal melanoma: epidemiology, etiology, and treatment of primary disease

Benjamin A Krantz, Nikita Dave, Kimberly M Komatsubara, Brian P Marr, Richard D Carvajal, Benjamin A Krantz, Nikita Dave, Kimberly M Komatsubara, Brian P Marr, Richard D Carvajal

Abstract

Uveal melanoma (UM) is the most common intraocular malignancy and arises from melanocytes in the iris, ciliary body, or choroid. Early diagnosis and local treatment is crucial, as survival correlates with primary tumor size. However, approximately 50% of patients will develop metastatic disease with 6-12 months' survival from metastatic diagnosis. Genomic analyses have led to the development of gene-expression profiles that effectively predict metastatic progression; unfortunately, no adjuvant therapy has been shown to prolong survival to date. New insights into the molecular biology of UM have found frequent activating mutations in genes encoding for the G-protein α-subunit, GNAQ and GNA11, and improved understanding of the downstream signaling pathways MAPK, PI3K/Akt, and Hippo have afforded an array of new targets for treatment of this disease. Studies are under way with rationally developed regimens targeting these pathways, and novel agents are under development. We review the diagnosis, management, and surveillance of primary UM and the adjuvant therapy trials under way.

Keywords: GNA11; GNAQ; MAP kinase; MEK; ocular melanoma; uveal melanoma.

Conflict of interest statement

Disclosure RDC serves a consultant and/or advisory board member for AstraZeneca, Aura Biosciences, Iconic Therapeutics, Janssen, Merck, Novartis, Rgenix, and Thomson Reuters. BPM serves as a consultant for Aura Biosciences. The other authors report no conflicts of interest in this work.

Figures

Figure 1
Figure 1
Signaling pathways in uveal melanoma. Abbreviations: PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol (3,4,5)-trisphosphate; Gα, G-protein alpha subunit; GDP, guanosine diphosphate; GPCR, g-protein coupled receptor; GTP, guanosine triphosphate; P, phosphate; RTK, Receptor tyrosine kinase.

References

    1. McLaughlin CC, Wu XC, Jemal A, Martin HJ, Roche LM, Chen VW. Incidence of noncutaneous melanomas in the U.S. Cancer. 2005;103(5):1000–1007.
    1. Singh AD, Topham A. Incidence of uveal melanoma in the United States: 1973–1997. Ophthalmology. 2003;110(5):956–961.
    1. Damato B. Progress in the management of patients with uveal melanoma: the 2012 Ashton Lecture. Eye (Lond) 2012;26(9):1157–1172.
    1. Damato EM, Damato BE. Detection and time to treatment of uveal melanoma in the United Kingdom: an evaluation of 2,384 patients. Ophthalmology. 2012;119(8):1582–1589.
    1. Andreoli MT, Mieler WF, Leiderman YI. Epidemiological trends in uveal melanoma. Br J Ophthalmol. 2015;99(11):1550–1553.
    1. Singh AD, Turell ME, Topham AK. Uveal melanoma: trends in incidence, treatment, and survival. Ophthalmology. 2011;118(9):1881–1885.
    1. Hu DN, Yu GP, McCormick SA, Schneider S, Finger PT. Population-based incidence of uveal melanoma in various races and ethnic groups. Am J Ophthalmol. 2005;140(4):612–617.
    1. Virgili G, Gatta G, Ciccolallo L, et al. Incidence of uveal melanoma in Europe. Ophthalmology. 2007;114(12):2309–2315.
    1. Park SJ, Oh CM, Kim BW, Woo SJ, Cho H, Park KH. Nationwide incidence of ocular melanoma in South Korea by using the National Cancer Registry Database (1999–2011) Invest Ophthalmol Vis Sci. 2015;56(8):4719–4724.
    1. Shields CL, Kaliki S, Livesey M, et al. Association of ocular and oculodermal melanocytosis with the rate of uveal melanoma metastasis: analysis of 7872 consecutive eyes. JAMA Ophthalmol. 2013;131(8):993–1003.
    1. Weis E, Shah CP, Lajous M, Shields JA, Shields CL. The association of cutaneous and iris nevi with uveal melanoma: a meta-analysis. Ophthalmology. 2009;116(3):536–543.e2.
    1. Harbour JW, Onken MD, Roberson ED, et al. Frequent mutation of BAP1 in metastasizing uveal melanomas. Science. 2010;330(6009):1410–1413.
    1. Gallagher RP, Elwood JM, Rootman J, et al. Risk factors for ocular melanoma: Western Canada Melanoma Study. J Natl Cancer Inst. 1985;74(4):775–778.
    1. Lains I, Bartosch C, Mondim V, et al. Second primary neoplasms in patients with uveal melanoma: a SEER database analysis. Am J Ophthalmol. 2016;165:54–64.
    1. Tucker MA, Shields JA, Hartge P, Augsburger J, Hoover RN, Fraumeni JF., Jr Sunlight exposure as risk factor for intraocular malignant melanoma. N Engl J Med. 1985;313(13):789–792.
    1. Shah CP, Weis E, Lajous M, Shields JA, Shields CL. Intermittent and chronic ultraviolet light exposure and uveal melanoma: a meta-analysis. Ophthalmology. 2005;112(9):1599–1607.
    1. de Lange MJ, Razzaq L, Versluis M, et al. Distribution of GNAQ and GNA11 mutation signatures in uveal melanoma points to a light dependent mutation mechanism. PLoS One. 2015;10(9):e0138002.
    1. Shields CL, Furuta M, Berman EL, et al. Choroidal nevus transformation into melanoma: analysis of 2514 consecutive cases. Arch Ophthalmol. 2009;127(8):981–987.
    1. Shields JA, Sanborn GE, Augsburger JJ. The differential diagnosis of malignant melanoma of the iris: a clinical study of 200 patients. Ophthalmology. 1983;90(6):716–720.
    1. Shields JA, Augsburger JJ, Brown GC, Stephens RF. The differential diagnosis of posterior uveal melanoma. Ophthalmology. 1980;87(6):518–522.
    1. Accuracy of diagnosis of choroidal melanomas in the Collaborative Ocular Melanoma Study: COMS report no. 1. Arch Ophthalmol. 1990;108(9):1268–1273. No authors listed.
    1. Shields JA, Shields CL, Ehya H, Eagle RC, Jr, De Potter P. Fine-needle aspiration biopsy of suspected intraocular tumors. Int Ophthalmol Clin. 1993;33(3):77–82.
    1. Char DH, Miller T. Accuracy of presumed uveal melanoma diagnosis before alternative therapy. Br J Ophthalmol. 1995;79(7):692–696.
    1. McCannel TA. Fine-needle aspiration biopsy in the management of choroidal melanoma. Curr Opin Ophthalmol. 2013;24(3):262–266.
    1. Rietschel P, Panageas KS, Hanlon C, Patel A, Abramson DH, Chapman PB. Variates of survival in metastatic uveal melanoma. J Clin Oncol. 2005;23(31):8076–8080.
    1. Diener-West M, Reynolds SM, Agugliaro DJ, et al. Development of metastatic disease after enrollment in the COMS trials for treatment of choroidal melanoma: Collaborative Ocular Melanoma Study Group report no. 26. Arch Ophthalmol. 2005;123(12):1639–1643.
    1. Kujala E, Makitie T, Kivela T. Very long-term prognosis of patients with malignant uveal melanoma. Invest Ophthalmol Vis Sci. 2003;44(11):4651–4659.
    1. Kuk D, Shoushtari AN, Barker CA, et al. Prognosis of mucosal, uveal, acral, nonacral cutaneous, and unknown primary melanoma from the time of first metastasis. Oncologist. 2016;21(7):848–854.
    1. Shields CL, Kaliki S, Furuta M, Fulco E, Alarcon C, Shields JA. American Joint Committee on Cancer classification of posterior uveal melanoma (tumor size category) predicts prognosis in 7,731 patients. Ophthalmology. 2013;120(10):2066–2071.
    1. Shields CL, Kaliki S, Furuta M, Fulco E, Alarcon C, Shields JA. American Joint Committee on Cancer classification of uveal melanoma (anatomic stage) predicts prognosis in 7,731 patients: the 2013 Zimmerman Lecture. Ophthalmology. 2015;122(6):1180–1186.
    1. Versluis M, de Lange MJ, van Pelt SI, et al. Digital PCR validates 8q dosage as prognostic tool in uveal melanoma. PLoS One. 2015;10(3):e0116371.
    1. Ewens KG, Kanetsky PA, Richards-Yutz J, et al. Genomic profile of 320 uveal melanoma cases: chromosome 8p-loss and metastatic outcome. Invest Ophthalmol Vis Sci. 2013;54(8):5721–5729.
    1. Onken MD, Worley LA, Char DH, et al. Collaborative Ocular Oncology Group report number 1: prospective validation of a multi-gene prognostic assay in uveal melanoma. Ophthalmology. 2012;119(8):1596–1603.
    1. Field MG, Decatur CL, Kurtenbach S, et al. PRAME as an independent biomarker for metastasis in uveal melanoma. Clin Cancer Res. 2016;22(5):1234–1242.
    1. Cancer Genome Atlas Network Genomic classification of cutaneous melanoma. Cell. 2015;161(7):1681–1696.
    1. Kalinec G, Nazarali AJ, Hermouet S, Xu N, Gutkind JS. Mutated α subunit of the Gq protein induces malignant transformation in NIH 3T3 cells. Mol Cell Biol. 1992;12(10):4687–4693.
    1. Van Raamsdonk CD, Bezrookove V, Green G, et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457(7229):599–602.
    1. Van Raamsdonk CD, Griewank KG, Crosby MB, et al. Mutations in GNA11 in uveal melanoma. N Engl J Med. 2010;363(23):2191–2199.
    1. Piperno-Neumann S, Kapiteijn E, Larkin JM, et al. Landscape of genetic alterations in patients with metastatic uveal melanoma. J Clin Oncol. 2014;32(5 Suppl):9043.
    1. Griewank KG, van de Nes J, Schilling B, et al. Genetic and clinicopathologic analysis of metastatic uveal melanoma. Mod Pathol. 2014;27(2):175–183.
    1. Xu X, Wei WB, Li B, Gao F, Zhang Z, Jonas JB. Oncogenic GNAQ and GNA11 mutations in uveal melanoma in Chinese. PLoS One. 2014;9(10):e109699.
    1. Johansson P, Aoude LG, Wadt K, et al. Deep sequencing of uveal melanoma identifies a recurrent mutation in PLCB4. Oncotarget. 2016;7(4):4624–4631.
    1. Moore AR, Ceraudo E, Sher JJ, et al. Recurrent activating mutations of G-protein-coupled receptor CYSLTR2 in uveal melanoma. Nat Genet. 2016;48(6):675–680.
    1. Shoushtari AN, Carvajal RD. GNAQ and GNA11 mutations in uveal melanoma. Melanoma Res. 2014;24(6):525–534.
    1. Lee CH, Park D, Wu D, Rhee SG, Simon MI. Members of the Gq α subunit gene family activate phospholipase C β isozymes. J Biol Chem. 1992;267(23):16044–16047.
    1. Wu DQ, Lee CH, Rhee SG, Simon MI. Activation of phospholipase C by the α subunits of the Gq and G11 proteins in transfected Cos-7 cells. J Biol Chem. 1992;267(3):1811–1817.
    1. Rozengurt E. Mitogenic signaling pathways induced by G protein-coupled receptors. J Cell Physiol. 2007;213(3):589–602.
    1. Cobb MH, Goldsmith EJ. How MAP kinases are regulated. J Biol Chem. 1995;270(25):14843–14846.
    1. Populo H, Soares P, Rocha AS, Silva P, Lopes JM. Evaluation of the mTOR pathway in ocular (uvea and conjunctiva) melanoma. Melanoma Res. 2010;20(2):107–117.
    1. Babchia N, Calipel A, Mouriaux F, Faussat AM, Mascarelli F. The PI3K/Akt and mTOR/P70S6K signaling pathways in human uveal melanoma cells: interaction with B-Raf/ERK. Invest Ophthalmol Vis Sci. 2010;51(1):421–429.
    1. Khalili JS, Yu X, Wang J, et al. Combination small molecule MEK and PI3K inhibition enhances uveal melanoma cell death in a mutant GNAQ- and GNA11-dependent manner. Clin Cancer Res. 2012;18(16):4345–4355.
    1. Ho AL, Musi E, Ambrosini G, et al. Impact of combined mTOR and MEK inhibition in uveal melanoma is driven by tumor genotype. PLoS One. 2012;7(7):e40439.
    1. Overholtzer M, Zhang J, Smolen GA, et al. Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc Natl Acad Sci U S A. 2006;103(33):12405–12410.
    1. Zhao B, Ye X, Yu J, et al. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev. 2008;22(14):1962–1971.
    1. Hao Y, Chun A, Cheung K, Rashidi B, Yang X. Tumor suppressor LATS1 is a negative regulator of oncogene YAP. J Biol Chem. 2008;283(9):5496–5509.
    1. Feng X, Degese MS, Iglesias-Bartolome R, et al. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated Rho GTPase signaling circuitry. Cancer Cell. 2014;25(6):831–845.
    1. Yavuzyigitoglu S, Koopmans AE, Verdijk RM, et al. Uveal melanomas with SF3B1 mutations: a distinct subclass associated with late-onset metastases. Ophthalmology. 2016;123(5):1118–1128.
    1. Harbour JW, Roberson ED, Anbunathan H, Onken MD, Worley LA, Bowcock AM. Recurrent mutations at codon 625 of the splicing factor SF3B1 in uveal melanoma. Nat Genet. 2013;45(2):133–135.
    1. Martin M, Masshöfer L, Temming P, et al. Exome sequencing identifies recurrent somatic mutations in EIF1AX and SF3B1 in uveal melanoma with disomy 3. Nat Genet. 2013;45(8):933–936.
    1. Maat W, van der Velden PA, Out-Luiting C, et al. Epigenetic inactivation of RASSF1a in uveal melanoma. Invest Ophthalmol Vis Sci. 2007;48(2):486–490.
    1. Merhavi E, Cohen Y, Avraham BC, et al. Promoter methylation status of multiple genes in uveal melanoma. Invest Ophthalmol Vis Sci. 2007;48(10):4403–4406.
    1. Rajaii F, Asnaghi L, Enke R, Merbs SL, Handa JT, Eberhart CG. The demethylating agent 5-Aza reduces the growth, invasiveness, and clonogenicity of uveal and cutaneous melanoma. Invest Ophthalmol Vis Sci. 2014;55(10):6178–6186.
    1. Venza M, Visalli M, Catalano T, et al. DNA methylation-induced E-cadherin silencing is correlated with the clinicopathological features of melanoma. Oncol Rep. 2016;35(4):2451–2460.
    1. Eldh M, Bagge RO, Lasser C, et al. MicroRNA in exosomes isolated directly from the liver circulation in patients with metastatic uveal melanoma. BMC Cancer. 2014;14:962.
    1. Chen X, He D, Da Dong X, et al. MicroRNA-124a is epigenetically regulated and acts as a tumor suppressor by controlling multiple targets in uveal melanoma miR-124a is a tumor suppressor in uveal melanoma. Invest Ophthalmol Vis Sci. 2013;54(3):2248–2256.
    1. Chen X, Wang J, Shen H, et al. Epigenetics, microRNAs, and carcinogenesis: functional role of microRNA-137 in uveal melanoma. Invest Ophthalmol Vis Sci. 2011;52(3):1193–1199.
    1. Dong F, Lou D. MicroRNA-34b/c suppresses uveal melanoma cell proliferation and migration through multiple targets. Mol Vis. 2012;18:537–546.
    1. Wen X, Lu L, He Z, Fan X. Orchestrating epigenetic roles targeting ocular tumors. Onco Targets Ther. 2016;9:1001–1009.
    1. Collaborative Ocular Melanoma Study Group The COMS randomized trial of iodine 125 brachytherapy for choroidal melanoma: V. Twelve-year mortality rates and prognostic factors: COMS report no. 28. Arch Ophthalmol. 2006;124(12):1684–1693.
    1. Tarmann L, Wackernagel W, Avian A, et al. Ruthenium-106 plaque brachytherapy for uveal melanoma. Br J Ophthalmol. 2015;99(12):1644–1649.
    1. American Brachytherapy Society – Ophthalmic Oncology Task Force The American Brachytherapy Society consensus guidelines for plaque brachytherapy of uveal melanoma and retinoblastoma. Brachytherapy. 2014;13(1):1–14.
    1. Puusaari I, Damato B, Kivelä T. Transscleral local resection versus iodine brachytherapy for uveal melanomas that are large because of tumour height. Graefes Arch Clin Exp Ophthalmol. 2007;245(4):522–533.
    1. Finger PT, Chin KJ, Duvall G. Palladium-103 ophthalmic plaque radiation therapy for choroidal melanoma: 400 treated patients. Ophthalmology. 2009;116(4):790–796.e1.
    1. Caminal J, Padrón-Pérez N, Arias L, et al. Transscleral resection without hypotensive anaesthesia vs iodine-125 plaque brachytherapy in the treatment of choroidal melanoma. Eye (Lond) 2016;30(6):833–842.
    1. Bechrakis NE, Bornfeld N, Zöller I, Foerster MH. Iodine 125 plaque brachytherapy versus transscleral tumor resection in the treatment of large uveal melanomas. Ophthalmology. 2002;109(10):1855–1861.
    1. Shah SU, Shields CL, Bianciotto CG, et al. Intravitreal bevacizumab at 4-month intervals for prevention of macular edema after plaque radiotherapy of uveal melanoma. Ophthalmology. 2014;121(1):269–275.
    1. Mishra KK, Quivey JM, Daftari IK, et al. Long-term results of the UCSF-LBNL randomized trial: charged particle with helium ion versus iodine-125 plaque therapy for choroidal and ciliary body melanoma. Int J Radiat Oncol Biol Phys. 2015;92(2):376–383.
    1. Bensoussan E, Thariat J, Maschi C, et al. Outcomes after proton beam therapy for large choroidal melanomas in 492 patients. Am J Ophthalmol. 2016;165:78–87.
    1. Seibel I, Cordini D, Rehak M, et al. Local recurrence after primary proton beam therapy in uveal melanoma: risk factors, retreatment approaches, and outcome. Am J Ophthalmol. 2015;160(4):628–636.
    1. Hawkins BS. The Collaborative Ocular Melanoma Study (COMS) randomized trial of pre-enucleation radiation of large choroidal melanoma: IV. Ten-year mortality findings and prognostic factors. COMS report number 24. Am J Ophthalmol. 2004;138(6):936–951.
    1. Melia M, Moy CS, Reynolds SM, et al. Quality of life after iodine 125 brachytherapy vs enucleation for choroidal melanoma: 5-year results from the Collaborative Ocular Melanoma Study: COMS QOLS report no. 3. Arch Ophthalmol. 2006;124(2):226–238.
    1. Willerding GD, Cordini D, Moser L, Krause L, Foerster MH, Bechrakis NE. Neoadjuvant proton beam irradiation followed by transscleral resection of uveal melanoma in 106 cases. Br J Ophthalmol. 2016;100(4):463–467.
    1. Kivela T, Puusaari I, Damato B. Transscleral resection versus iodine brachytherapy for choroidal malignant melanomas 6 millimeters or more in thickness: a matched case-control study. Ophthalmology. 2003;110(11):2235–2244.
    1. Bechrakis NE, Petousis V, Willerding G, et al. Ten-year results of transscleral resection of large uveal melanomas: local tumour control and metastatic rate. Br J Ophthalmol. 2010;94(4):460–466.
    1. Badiyan SN, Rao RC, Apicelli AJ, et al. Outcomes of iodine-125 plaque brachytherapy for uveal melanoma with intraoperative ultrasonography and supplemental transpupillary thermotherapy. Int J Radiat Oncol Biol Phys. 2014;88(4):801–805.
    1. Saakian SV, Val’skii VV, Semenova EA, Amirian AG. Transpupillary thermotherapy in the treatment of recurrent and residual choroidal melanomas: preliminary results. Vestn Oftalmol. 2009;125(6):11–15. Russian.
    1. Harbour JW, Meredith TA, Thompson PA, Gordon ME. Transpupillary thermotherapy versus plaque radiotherapy for suspected choroidal melanomas. Ophthalmology. 2003;110(11):2207–2215.
    1. Turcotte S, Bergeron D, Rousseau AP, Mouriaux F. Primary transpupillary thermotherapy for choroidal indeterminate melanocytic lesions. Can J Ophthalmol. 2014;49(5):464–467.
    1. Mashayekhi A, Shields CL, Rishi P, et al. Primary transpupillary thermotherapy for choroidal melanoma in 391 cases: importance of risk factors in tumor control. Ophthalmology. 2015;122(3):600–609.
    1. Yarovoy AA, Magaramov DA, Bulgakova ES. The comparison of ruthenium brachytherapy and simultaneous transpupillary thermotherapy of choroidal melanoma with brachytherapy alone. Brachytherapy. 2012;11(3):224–229.
    1. Walker TM, Van Ginkel PR, Gee RL, et al. Expression of angiogenic factors Cyr61 and tissue factor in uveal melanoma. Arch Ophthalmol. 2002;120(12):1719–1725.
    1. Kines RC, Cerio RJ, Roberts JN, et al. Human papillomavirus capsids preferentially bind and infect tumor cells. Int J Cancer. 2016;138(4):901–911.
    1. Business Wire FDA grants orphan drug designation to Aura Biosciences’ novel treatment for uveal melanoma. 2015. [Accessed November 26, 2016]. Available from: .
    1. Torres V, Triozzi P, Eng C, et al. Circulating tumor cells in uveal melanoma. Future Oncol. 2011;7(1):101–109.
    1. Desjardins L, Dorval T, Levy C, et al. Etude randomisée de chimiothérapie adjuvante par le deticène dans le mélanome choroïdien [Randomized study of adjuvant chemotherapy with dacarbazine in choroidal melanoma] Ophtalmologie. 1998;12(3):168–173. French.
    1. Lane AM, Egan KM, Harmon D, Holbrook A, Munzenrider JE, Gragoudas ES. Adjuvant interferon therapy for patients with uveal melanoma at high risk of metastasis. Ophthalmology. 2009;116(11):2206–2212.
    1. Richtig E, Langmann G, Schlemmer G, et al. Safety and efficacy of interferon alfaα-2b in the adjuvant treatment of uveal melanoma. Ophthalmologe. 2006;103(6):506–511. German.
    1. National Comprehensive Cancer Network . NCCN Guidelines: Melanoma Version 3.2016. Fort Washington (PA): NCCN; 2016.
    1. Leyvraz S, Spataro V, Bauer J, et al. Treatment of ocular melanoma metastatic to the liver by hepatic arterial chemotherapy. J Clin Oncol. 1997;15(7):2589–2595.
    1. Voelter V, Schalenbourg A, Pampallona S, et al. Adjuvant intra-arterial hepatic fotemustine for high-risk uveal melanoma patients. Melanoma Res. 2008;18(3):220–224.
    1. Leyvraz S, Piperno-Neumann S, Suciu S, et al. Hepatic intra-arterial versus intravenous fotemustine in patients with liver metastases from uveal melanoma (EORTC 18021): a multicentric randomized trial. Ann Oncol. 2014;25(3):742–746.
    1. Eder JP, Vande Woude GF, Boerner SA, LoRusso PM. Novel therapeutic inhibitors of the c-Met signaling pathway in cancer. Clin Cancer Res. 2009;15(7):2207–2214.
    1. Liang J, Wu YL, Chen BJ, Zhang W, Tanaka Y, Sugiyama H. The c-Kit receptor-mediated signal transduction and tumor-related diseases. Int J Biol Sci. 2013;9(5):435–443.
    1. Sato T, Han F, Yamamoto A. The biology and management of uveal melanoma. Curr Oncol Rep. 2008;10(5):431–438.
    1. Surriga O, Rajasekhar VK, Ambrosini G, Dogan Y, Huang R, Schwartz GK. Crizotinib, a c-Met inhibitor, prevents metastasis in a metastatic uveal melanoma model. Mol Cancer Ther. 2013;12(12):2817–2826.
    1. Valsecchi ME, Orloff MM, Chervoneva I, et al. Potential survival benefit of adjuvant sunitinib in high risk uveal melanoma. J Clin Oncol. 2015;33(Suppl):e20046.
    1. Landreville S, Agapova OA, Matatall KA, et al. Histone deacetylase inhibitors induce growth arrest and differentiation in uveal melanoma. Clin Cancer Res. 2012;18(2):408–416.
    1. Kaiserman I, Amer R, Pe’er J. Liver function tests in metastatic uveal melanoma. Am J Ophthalmol. 2004;137(2):236–243.
    1. Koutsandrea C, Moschos MM, Dimissianos M, Georgopoulos G, Ladas I, Apostolopoulos M. Metastasis rates and sites after treatment for choroidal melanoma by proton beam irradiation or by enucleation. Clin Ophthalmol. 2008;2(4):989–995.
    1. Francis JH, Patel SP, Gombos DS, Carvajal RD. Surveillance options for patients with uveal melanoma following definitive management. Am Soc Clin Oncol Educ Book. 2013:382–387.
    1. Francken AB, Fulham MJ, Millward MJ, Thompson JF. Detection of metastatic disease in patients with uveal melanoma using positron emission tomography. Eur J Surg Oncol. 2006;32(7):780–784.
    1. Chattopadhyay C, Kim DW, Gombos DS, et al. Uveal melanoma: from diagnosis to treatment and the science in between. Cancer. 2016;122(15):2299–2312.
    1. Piperno-Neumann S, Servois V, Mariani P, et al. Prospective study of surveillance testing for metastasis in 100 high-risk uveal melanoma patients. J Fr Ophtalmol. 2015;38(6):526–534.
    1. Marshall E, Romaniuk C, Ghaneh P, et al. MRI in the detection of hepatic metastases from high-risk uveal melanoma: a prospective study in 188 patients. Br J Ophthalmol. 2013;97(2):159–163.
    1. Wen JC, Sai V, Straatsma BR, McCannel TA. Radiation-related cancer risk associated with surveillance imaging for metastasis from choroidal melanoma. JAMA Ophthalmol. 2013;131(1):56–61.
    1. Carvajal RD, Schwartz GK, Tezel T, Marr B, Francis JH, Nathan PD. Metastatic disease from uveal melanoma: treatment options and future prospects. Br J Ophthalmol. 2016 Aug 29; Epub.
    1. Guan X, Wang H, Ma F, Qian H, Yi Z, Xu B. The efficacy and safety of programmed cell death 1 and programmed cell death 1 ligand inhibitors for advanced melanoma: a meta-analysis of clinical trials following the PRISMA guidelines. Medicine (Baltimore) 2016;95(11):e3134.
    1. Bossi G, Buisson S, Oates J, Jakobsen BK, Hassan NJ. ImmTAC- redirected tumour cell killing induces and potentiates antigen cross- presentation by dendritic cells. Cancer Immunol Immunother. 2014;63(5):437–448.
    1. Marquez-Rodas I, Cerezuela P, Soria A, et al. Immune checkpoint inhibitors: therapeutic advances in melanoma. Ann Transl Med. 2015;3(18):267.
    1. Middleton MR, Steven NM, Evans TJ, et al. Safety, pharmacokinetics and efficacy of IMCgp100, a first-in-class soluble TCR-antiCD3 bispecific T cell redirector with solid tumour activity: results from the FIH study in melanoma. J Clin Oncol. 2016;34(Suppl):3016.

Source: PubMed

3
订阅